EP3386506B1 - Modulators of kv3 channels to treat pain - Google Patents

Modulators of kv3 channels to treat pain Download PDF

Info

Publication number
EP3386506B1
EP3386506B1 EP16813020.1A EP16813020A EP3386506B1 EP 3386506 B1 EP3386506 B1 EP 3386506B1 EP 16813020 A EP16813020 A EP 16813020A EP 3386506 B1 EP3386506 B1 EP 3386506B1
Authority
EP
European Patent Office
Prior art keywords
oxy
methyl
alkyl
pain
ethyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP16813020.1A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP3386506A1 (en
Inventor
Charles Large
Giuseppe Alvaro
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Autifony Therapeutics Ltd
Original Assignee
Autifony Therapeutics Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Autifony Therapeutics Ltd filed Critical Autifony Therapeutics Ltd
Publication of EP3386506A1 publication Critical patent/EP3386506A1/en
Application granted granted Critical
Publication of EP3386506B1 publication Critical patent/EP3386506B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • A61P29/02Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID] without antiinflammatory effect
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C43/00Ethers; Compounds having groups, groups or groups
    • C07C43/30Compounds having groups
    • C07C43/315Compounds having groups containing oxygen atoms singly bound to carbon atoms not being acetal carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/94Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom spiro-condensed with carbocyclic rings or ring systems, e.g. griseofulvins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings

Definitions

  • This invention relates to compounds and pharmaceutical compositions containing such compounds for use in the prophylaxis or treatment of pain, and to related methods and uses.
  • the Kv3 voltage-gated potassium channel family includes four members, Kv3.1, Kv3.2, Kv3.3, and Kv3.4. Genes for each of these subtypes can generate multiple isoforms by alternative splicing, producing versions with different C-terminal domains. Thirteen isoforms have been identified in mammals to date, but the currents expressed by these variants appear similar (Rudy et al., 2001). Kv3 channels are activated by depolarisation of the plasma membrane to voltages more positive than -20mV; furthermore, the channels deactivate rapidly upon repolarisation of the membrane. These biophysical properties ensure that the channels open towards the peak of the depolarising phase of the neuronal action potential to initiate repolarisation.
  • Kv3.1-Kv3.3 subtypes are predominant in the CNS, whereas Kv3.4 channels are found predominantly in skeletal muscle and sympathetic neurons (Weiser et al., 1994).
  • Kv3.1-Kv3.3 channel subtypes are differentially expressed by sub-classes of interneurons in cortical and hippocampal brain areas (e.g.
  • Kv3 channels are important determinants of the function of the cerebellum, a region of the brain important for motor control (Joho et al., 2009). Characterisation of mice in which one or more of the Kv3 subtypes has been deleted shows that the absence of Kv3.1 gives rise to increased locomotor activity, altered electroencephalographic activity, and a fragmented sleep pattern (Joho et al., 1999). The deletion of Kv3.2 leads to a reduction in seizure threshold and altered cortical electroencephalographic activity (Lau et al., 2000). Deletion of Kv3.3 is associated with mild ataxia and motor deficits (McMahon et al., 2004).
  • Double deletion of Kv3.1 and Kv3.3 gives rise to a severe phenotype characterised by spontaneous seizures, ataxia, and an increased sensitivity to the effects of ethanol (Espinosa et al., 2001; Espinosa et al., 2008).
  • TAA Tetraethylammonium
  • BDS blood-depressing substance
  • Patent applications WO2011/069951 , WO2012/076877 , WO2012/168710 , WO2013/175215 , WO2013/083994 and WO2013/182850 disclose compounds which are modulators of Kv3 channels, specifically Kv3.1, Kv3.2 and Kv3.3. Use of such compounds in certain diseases and disorders requiring modulation of Kv3 channels are disclosed in patent applications WO2013/182851 and WO2013/175211 .
  • pain can be grouped in to acute pain and chronic pain.
  • Acute pain is defined as pain that is self-limited and generally requires treatment for no more than up to a few weeks, for example postoperative or acute musculoskeletal pain, such as fractures (US Food and Drug Administration, 2014).
  • Chronic pain can be defined either as pain persisting for longer than 1 month beyond resolution of the initial trauma, or pain persisting beyond three months. There is often no clear cause of chronic pain, and a multitude of other health problems such as fatigue, depression, insomnia, mood changes and reduction in movement, often accompany chronic pain.
  • Chronic pain can be sub-divided in to the following groups: neuropathic pain, chronic musculoskeletal pain and miscellaneous chronic pain.
  • Neuropathic pain usually accompanies tissue injury and is initiated or caused by damage to the nervous system (peripheral nervous system and/or central nervous system), such as amputation, stroke, diabetes, or multiple sclerosis.
  • Chronic musculoskeletal pain can be a symptom of diseases such as osteoarthritis and chronic lower back pain and can occur following damage to muscle tissue as well as trauma to an area, for example, fractures, sprains and dislocation.
  • Miscellaneous chronic pain encompasses all other types of long term pain and includes non-neuropathic pain conditions such as cancer pain and fibromyalgia as well as headaches and tendinitis.
  • Voltage-gated ion channels have been important targets for the management of specific pain indications, in particular neuropathic pain states. Furthermore, genetic mutations in specific ion channels have been linked to some chronic pain disorders (Bennett et al., 2014). Examples of voltage-gated ion channels that are being explored as pharmaceutical targets include:
  • Drugs targeting hyperexcitability such as sodium channel blockers (e.g. CNV1014802, lamotrigine, carbamazepine, and local anaesthetics), Kv7 positive modulators (e.g. flupertine and retigabine), and N-type calcium channel modulators (e.g. gabapentin, which interacts with the ⁇ 2 ⁇ subunit of the N-type calcium channel, and ziconitide, derived from a cone snail toxin) show efficacy in models of inflammatory and/or neuropathic pain.
  • sodium channel blockers e.g. CNV1014802, lamotrigine, carbamazepine, and local anaesthetics
  • Kv7 positive modulators e.g. flupertine and retigabine
  • N-type calcium channel modulators e.g. gabapentin, which interacts with the ⁇ 2 ⁇ subunit of the N-type calcium channel, and ziconitide, derived from a cone snail toxin
  • Improving the pharmacological management of pain is focused on mechanisms that can deliver good efficacy with a reduced side-effect burden, reduced tolerance or tachyphylaxis, and reduced abuse liability and/or risk of dependence.
  • Kv3.4 channels have become a target of interest for the treatment of chronic pain.
  • Kv3.4 channels are expressed on neurons of the dorsal root ganglia (Ritter et al., 2012; Chien et al., 2007), where they are predominantly expressed on sensory C-fibres (Chien et al., 2007).
  • Kv3 channels are also expressed by specific subsets of neurons in the spinal cord. Specifically, Kv3.1b (Deuchars et al., 2001; Brooke et al., 2002), Kv3.3 (Brooke et al., 2006), and Kv3.4 subunits (Brooke et al., 2004) have been identified in rodent spinal cord, although not always in association with circuits involved with sensory processing.
  • Kv3.4 channel inactivation could be influenced by protein kinase C-dependent phosphorylation of the channels, and that this physiological mechanism might allow DRG neurons to alter their firing characteristics in response to painful stimuli (Ritter et al., 2012).
  • These studies suggest a causal relationship between the emergence of mechanical allodynia and reduced Kv3.4 channel expression or function.
  • No evaluation of Kv3.1, Kv3.2, or Kv3.3 expression in SC or DRG neurons was conducted in any of these studies, and expression of these three subtypes has not been explicitly demonstrated on DRG neurons (although as mentioned above, they are abundant within specific regions of the spinal cord).
  • modulation of Kv3.1 and/or Kv3.2 and/or Kv3.3 channels is linked to the processing of pain and pain control. Therefore, modulation of Kv3.1 and/or Kv3.2 and/or Kv3.3 represents a new approach for the prophylaxis or treatment of pain.
  • the present invention provides a modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 channels (referred to herein as "Kv3.1/Kv3.2/Kv3.3” or "Kv3.1 and/or Kv3.2 and/or Kv3.3”) for use in the prophylaxis or treatment of pain.
  • the modulator is a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate thereof: wherein:
  • the compounds of formula (I) may be used as medicaments, in particular for the prophylaxis or treatment of pain, such as neuropathic or inflammatory pain.
  • a method of identifying that a compound is of use in the prophylaxis or treatment of pain comprising the step of determining that the compound is a modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 channels.
  • the present invention provides a modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 channels for use in the prophylaxis or treatment of pain.
  • the modulator is a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate thereof: wherein:
  • the compounds of formula (I) may be used as medicaments, in particular for the prophylaxis or treatment of pain, such as neuropathic or inflammatory pain.
  • the modulator is a compound of formula (IA): wherein R 1 , R 2 , R 3 , R 13 , R 14 , A, X, Y, R 4 and R 5 are as defined above for compounds for formula (I).
  • the modulator is a compound of formula (IB): wherein R 1 , R 2 , R 3 , R 13 , R 14 , A, X, Y and R 4 are as defined above for compounds for formula (I).
  • the modulator is a compound of formula (IC): wherein R 1 , R 2 , R 3 , R 13 , R 14 , A, X, Y, R 4 and R 5 are as defined above for compounds for formula (I).
  • the modulator is a compound of formula (ID): wherein R 1 , R 2 , R 3 , R 13 , R 14 , A, X, Y and R 4 are as defined above for compounds for formula (I).
  • the modulator is a compound of formula (IE): wherein R 16 and R 17 , X, Y, R 4 and R 5 are as defined above for compounds for formula (I).
  • the modulator is a compound of formula (IF): wherein R 22 , R 23 , R 24 , R 25 , R 26 , X, Y and R 4 are as defined above for compounds for formula (I).
  • R 1 is H, C 1-4 alkyl, halo or haloC 1-4 alkyl. In another embodiment of the invention R 1 is H or methyl. In one embodiment of the invention R 1 is H. In another embodiment of the invention R 1 is C 1-4 alkyl, in particular methyl.
  • W is group (Wa)
  • W is group (Wb)
  • R 1 is H or methyl.
  • R 1 is positioned at the para position of the phenyl ring, as illustrated below:
  • R 2 is H, C 1-4 alkyl, C 3-5 spiro carbocyclyl, or haloC 1-4 alkyl.
  • R 2 is C 1-4 alkyl, in particular methyl, ethyl, isopropyl, tert-butyl or cyclopropyl, especially methyl, ethyl, isopropyl or tert-butyl.
  • R 2 is C 3-5 spiro carbocyclyl.
  • R 2 is C 3 spiro carbocyclyl.
  • R 2 is C 4 spiro carbocyclyl.
  • R 2 is C 5 spiro carbocyclyl.
  • R 2 is haloC 1-4 alkyl, in particular trifluoromethyl or 2,2,2-trifluoroethyl.
  • R 2 is halo, in particular fluoro.
  • R 2 is H.
  • R 3 is H, C 1-4 alkyl, haloC 1-4 alkyl or halo.
  • R 3 is H, C 1-4 alkyl, or haloC 1-4 alkyl.
  • R 3 is H or C 1-4 alkyl.
  • R 3 is H.
  • R 3 is C 1-4 alkyl, in particular methyl, ethyl, isopropyl, tert-butyl or cyclopropyl, especially methyl, ethyl, isopropyl or tert-butyl, such as methyl or ethyl.
  • R 3 is haloC 1-4 alkyl, in particular trifluoromethyl or 2,2,2-trifluoroethyl.
  • R 3 is halo, in particular fluoro.
  • R 3 is H, methyl or trifluoromethyl.
  • R 2 may be H, C 1-4 alkyl, haloC 1-4 alkyl or C 3-5 spiro carbocyclyl and R 3 may be H, C 1-4 alkyl, or haloC 1-4 alkyl.
  • R 2 may be methyl, ethyl, isopropyl, tert-butyl, cyclopropyl, C 3-5 spiro carbocyclyl, trifluoromethyl or 2,2,2-trifluoroethyl and R 3 may be H, methyl, ethyl or trifluoromethyl.
  • R 3 is H and R 2 is H, methyl, ethyl, isopropyl or C 3-4 spiro carbocyclyl.
  • R 3 and R 2 are both fluoro (such as attached to the same ring carbon atom).
  • R 2 is C 1-4 alkyl and R 3 is H, for example R 2 is methyl, ethyl, tert-butyl or cyclopropyl.
  • R 2 is C 1-4 alkyl and R 3 is C 1-4 alkyl, for example R 2 is methyl and R 3 is methyl, R 2 is ethyl and R 3 is ethyl or R 2 is methyl and R 3 is ethyl. In another embodiment of the invention R 2 is trifluoromethyl and R 3 is methyl.
  • R 2 and R 3 are attached to the same ring atom. In an alternative embodiment of the invention R 2 and R 3 are attached to different ring atoms.
  • R 13 is H, F or methyl. In one embodiment of the invention R 13 is H. In another embodiment of the invention R 13 is C 1-4 alkyl, in particular methyl. In a further embodiment of the invention R 13 is halo, in particular fluoro. In an additional embodiment of the invention R 13 is haloC 1-4 alkyl, such as trifluoromethyl.
  • R 13 may be absent. Consequently, in another embodiment of the invention R 13 is absent.
  • R 14 is H, F or methyl. In one embodiment of the invention R 14 is H. in another embodiment of the invention R 14 is C 1-4 alkyl, in particular methyl. In a further embodiment of the invention R 14 is halo, in particular fluoro. In an additional embodiment of the invention R 13 is haloC 1-4 alkyl, such as trifluoromethyl.
  • R 14 may be absent. Consequently, in another embodiment of the invention R 14 is absent.
  • R 13 and R 14 are attached to the same ring atom. In an alternative embodiment of the invention R 13 and R 14 are attached to different ring atoms.
  • R 2 , R 3 , R 13 and R 14 are each independently selected from H, C 1-4 alkyl, haloC 1-4 alkyl and halo, such as H, C 1-4 alkyl and haloC 1-4 alkyl.
  • R 2 , R 3 , R 13 and R 14 are each independently selected from H, F, methyl and trifluoromethyl.
  • A is a 5 or 6 membered saturated or unsaturated heterocycle, with at least one O atom; which heterocycle is optionally fused with a cyclopropyl group to form a tricycle when considered together with the phenyl.
  • A is a 5 membered saturated or unsaturated heterocycle, with at least one O atom; which heterocycle is optionally fused with a cyclopropyl group, a cyclobutyl group or a cyclopentyl group to form a tricycle when considered together with the phenyl.
  • A is a 6 membered saturated or unsaturated heterocycle, with at least one O atom; which heterocycle is optionally fused with a cyclopropyl group, a cyclobutyl group or a cyclopentyl group to form a tricycle when considered together with the phenyl.
  • A is a 5 membered saturated or unsaturated heterocycle with at least one O atom, which heterocycle is fused with a cyclopropyl group to form a tricycle when considered together with the phenyl.
  • A is a 6 membered saturated or unsaturated heterocycle with at least one O atom, which heterocycle is fused with a cyclopropyl group to form a tricycle when considered together with the phenyl.
  • A is a 5 membered saturated or unsaturated heterocycle with at least one O atom. In one embodiment of the invention A is a 6 membered saturated or unsaturated heterocycle with at least one O atom.
  • the ring A contains one heteroatom. In other embodiments of the invention the ring A contains two heteroatoms (e.g. two oxygen atoms, one oxygen atom and one nitrogen atom, or one oxygen atom and one sulphur atom), in particular two oxygen atoms or one oxygen atom and one nitrogen atom.
  • two heteroatoms e.g. two oxygen atoms, one oxygen atom and one nitrogen atom, or one oxygen atom and one sulphur atom
  • A is dihydrofuran, isoxazole, dihydropyran, 1,3-dioxolane, 1,3-oxazine or dihydropyran fused with a cyclopropyl group.
  • A is dihydrofuran. In one embodiment of the invention A is dihydropyran. In another embodiment of the invention A is dihydrofuran fused with a cyclopropyl group, a cyclobutyl group or a cyclopentyl group. In another embodiment of the invention A is dihydropyran fused with a cyclopropyl group, a cyclobutyl group or a cyclopentyl group. In a further embodiment the invention A is dihydrofuran fused with a cyclopropyl group. In still further embodiment the invention A is dihydropyran fused with a cyclopropyl group.
  • A is fused with a cyclopropyl group. In another embodiment A is fused with a cyclobutyl group. In a further embodiment of the invention A is fused with a cyclopentyl group. In one embodiment of the invention A is not fused with a cyclopropyl group, a cyclobutyl group or a cyclopentyl group.
  • A is dihydrofuran, dihydropyran, furan, pyran, oxazole, isoxazole, oxazine, dioxine or 1,3-dioxalane. In another embodiment A is dihydrofuran, dihydropyran or 1,3-dioxalane.
  • A is: wherein denotes a portion of the phenyl ring to which ring A is fused.
  • A is: wherein denotes a portion of the phenyl ring to which ring A is fused.
  • A is: wherein denotes a portion of the phenyl ring to which ring A is fused.
  • A contains a 5 membered heterocycle containing one oxygen atom
  • the heterocycle is dihydrofuran.
  • A is a 5 membered heterocycle containing one oxygen atom
  • the oxygen atom is located at the benzylic position relative to the phenyl ring.
  • W is group (Wa)
  • A is a 5 membered heterocycle containing one heteroatom, wherein the oxygen atom is located at the benzylic or para position relative to the phenyl ring.
  • W is group (Wb)
  • A is a 5 membered heterocycle containing one heteroatom, wherein the oxygen atom is located at the benzylic or meta position relative to the phenyl ring.
  • group (Wa) is:
  • group (Wa) is:
  • group (Wb) is:
  • (Wb) is:
  • group (Wb) is:
  • A contains a 6 membered heterocycle containing one oxygen atom
  • the heterocycle is dihydropyran.
  • W is group (Wa)
  • A is a 6 membered heterocycle containing one oxygen atom, wherein the oxygen atom is located at the para position relative to the phenyl ring.
  • W is group (Wb)
  • A contains a 6 membered heterocycle containing one oxygen atom, wherein the oxygen atom is located at the meta position relative to the phenyl ring.
  • group (Wa) is:
  • group (Wa) is:
  • group (Wb) is:
  • group (Wb) is:
  • group (Wb) is:
  • A is:
  • A is: wherein m and p denote the meta and para positions, respectively, of ring A relative to the phenyl ring.
  • A is selected from the group consisting of: wherein m and p denote the meta and para positions, respectively, of ring A relative to the phenyl ring.
  • W is group (Wb)
  • A is: wherein m and o denote the meta and ortho positions, respectively, of ring A relative to the phenyl ring.
  • W is group (Wb)
  • A is: wherein m and o denote the meta and ortho positions, respectively, of ring A relative to the phenyl ring.
  • W is the group (Wc):
  • R 16 is C 1-4 alkoxy. In another embodiment of the invention R 16 is methoxy. In one embodiment of the invention R 16 is C 1-4 alkyl. In another embodiment of the invention R 16 is methyl. In a further embodiment of the invention R 16 is ethyl. In a yet further embodiment of the invention R 16 is propyl. In a yet further embodiment of the invention R 16 is butyl. In one embodiment of the invention R 16 is halo. In another embodiment of the invention R 16 is chloro. In a further embodiment of the invention R 16 is fluoro. In one embodiment of the invention R 16 is halo-C 1-4 alkoxy. In another embodiment of the invention R 16 is trifluoromethoxy. In one embodiment of the invention R 16 is halo-C 1-4 alkyl. In another embodiment of the invention R 16 is trifluoromethyl. In one embodiment of the invention R 16 is cyano.
  • R 17 is H. In one embodiment of the invention R 17 is C 1-4 alkyl. In another embodiment of the invention R 17 is methyl. In one embodiment of the invention R 17 is halo. In another embodiment of the invention, R 17 is chloro. In a further embodiment of the invention R 17 is fluoro. In one embodiment of the invention R 17 is C 1-4 alkyl. In one embodiment of the invention R 17 is cyano.
  • R 16 is C 1-4 alkyl, C 1-4 alkoxy, or halo-C 1-4 alkoxy; R 17 is H, cyano or alkyl; X is N, Y is N or CR 15 , R 4 is C 1-4 alkyl, and R 5 is C 1-4 alkyl or H.
  • R 16 is propyl, butyl, methoxy, propoxy, or trifluoromethoxy; R 17 is H, cyano or methyl; X is N, Y is N or CR 15 , R 4 is ethyl, and R 5 is methyl or H.
  • one of R 16 and R 17 is in the para position and the remaining R 16 or R 17 is in the meta position. In one embodiment, one of R 16 and R 17 is in the para position and the remaining R 16 or R 17 is in the ortho position.
  • R 16 is C 1-4 alkoxy and R 17 is C 1-4 alkyl. In one embodiment of the invention R 16 is methoxy and R 17 is methyl. In one embodiment of the invention R 16 is C 1-4 alkoxy in the meta position and R 17 is C 1-4 alkyl in the para position. In a further embodiment of the invention R 16 is methoxy in the meta position, R 17 is methyl in the para position, R 4 is C 1-4 alkyl, R 5 is H, R 4 is in the R configuration.
  • R 16 is methoxy in the meta position
  • R 17 is methyl in the para position
  • X is N
  • Y is CH
  • R 4 is C 1-4 alkyl
  • R 5 is H and the absolute configuration of the stereogenic centre is R.
  • R 16 is methoxy in the meta position
  • R 17 is methyl in the para position
  • X is N
  • Y is CH
  • R 4 is ethyl
  • R 5 is H and the absolute configuration of the stereogenic centre is R.
  • W is the group (Wd):
  • R 22 , R 25 and R 26 are H.
  • R 23 is C 1-4 alkyl, Cl, CF 3 , O-C 1-4 alkyl, OCF 3 or N(CH 3 ) 2 , such as C 1-2 alkyl, CF 3 , O-C 1-2 alkyl or OCF 3 , in particular OCF 3 and R 24 is H, Cl, F, C 1-4 alkyl, O-C 1-4 alkyl, CN, OCF 3 , such as F, C 1-2 alkyl, CF 3 , O-C 1-2 alkyl or OCF 3 , in particular F or methyl and R 22 , R 25 and R 26 are H.
  • W is group (Wd)
  • R 22 to R 26 are H and one of R 22 to R 26 , in particular R 22 or R 23 , is other than H.
  • R 22 is other than H, suitably it is methyl.
  • R 23 is other than H, suitably it is OCF 3 .
  • R 4 is C 1-4 alkyl.
  • R 4 is methyl, ethyl, isopropyl or t-butyl.
  • R 4 is methyl.
  • R 4 is ethyl.
  • R 4 is propyl, such as isopropyl.
  • R 4 is butyl, such as t-butyl.
  • R 5 is H or C 1-4 alkyl. In one embodiment of the invention R 5 is H. In another embodiment of the invention R 4 is methyl, ethyl, isopropyl or t-butyl. In another embodiment of the invention R 4 is methyl. In a yet further embodiment of the invention R 4 is ethyl. In a yet further embodiment of the invention R 4 is propyl, such as isopropyl. In a yet further embodiment of the invention R 4 is butyl, such as t-butyl.
  • R 4 and R 5 together form a C 3 spiro carbocycle. In a second embodiment of the invention R 4 and R 5 together form a C 4 spiro carbocycle. In a further embodiment of the invention R 4 is methyl and R 5 is methyl. In an embodiment of particular interest, R 4 is ethyl and R 5 is methyl. In another embodiment, R 4 is ethyl and R 5 is ethyl. In an additional embodiment, R 4 is ethyl and R 5 is H.
  • R 4 and R 5 have the stereochemical arrangement:
  • R 4 is H.
  • R 4 is C 1-4 alkyl, in particular methyl, ethyl, isopropyl, tert-butyl or cyclopropyl.
  • R 4 is methyl.
  • R 4 is ethyl.
  • X is CH. In another embodiment of the invention X is N.
  • Y is CR 15 . In another embodiment of the invention Y is N. In a further embodiment of the invention Y is CR 15 , wherein R 15 is H. In a still further embodiment of the invention Y is CR 15 , wherein R 15 is C 1-4 alkyl, in particular methyl.
  • X is CH and Y is CR 15 , wherein R 15 is H. In another embodiment of the invention X is N and Y is CR 15 , wherein R 15 is H. In a further embodiment of the invention X is N and Y is CR 15 , wherein R 15 is methyl. In a further embodiment of the invention X is CH and Y is CR 15 , wherein R 15 is methyl. In a still further embodiment of the invention X is N and Y is N.
  • one embodiment of the invention provides a compound of formula (IFa): wherein:
  • R 22 is C 1-4 alkyl. In another embodiment R 22 is methyl. In a further embodiment R 22 is ethyl. In a yet further embodiment R 22 is propyl.
  • R 22 is Cl.
  • R 22 is F.
  • R 23 is H.
  • R 23 is C 1-4 alkyl. In another embodiment of the compounds of formula (IFa) R 23 is methyl.
  • R 23 is chloro
  • R 23 is methoxy. In another embodiment of the compounds of formula (IFa) R 23 is ethoxy.
  • R 23 is trifluoromethyl
  • R 23 is trifluoromethoxy
  • R 23 is N(CH 3 ) 2 .
  • R 24 is H.
  • R 24 is methyl
  • R 24 is chloro
  • R 24 is fluoro
  • R 25 is H.
  • R 25 is methyl
  • R 25 is chloro
  • R 25 is fluoro
  • R 26 is H.
  • R 26 is methyl
  • one embodiment of the invention provides a compound of formula (IFb): wherein:
  • R 4 is H.
  • R 4 is methyl
  • R 22 is H.
  • R 22 is methyl
  • R 23 is C 3 -C 4 alkyl. In another embodiment of the compounds of formula (IFb) R 23 is propyl.
  • R 23 is methyl
  • R 23 is OC 2 -C 4 alkyl. In another embodiment of the compounds of formula (IFb) R 23 is ethoxy.
  • R 24 is H.
  • R 25 is H.
  • R 26 is H.
  • R 15 is H.
  • R 15 is methyl
  • the compound of formula (I) may contain a W group corresponding to one of the following phenol groups:
  • the compound of formula (I) contains a (Wa) group corresponding to one of the following phenol groups:
  • the compound of formula (I) contains a (Wb) group corresponding to one of the following phenol groups:
  • the compound of formula (I) may contain a (Wb) group corresponding to one of the following phenol groups:
  • the compound of formula (I) is not a pharmaceutically acceptable salt (or not any salt).
  • the compound of formula (I) is not a solvate.
  • enantiomer 1, enantiomer 2, diastereomer 1 and diastereomer 2 refer to the particular enantiomer or disasteriomers named accordingly and described in the original disclosures of these compounds (see WO2011/069951 , WO2012/076877 , WO2012/168710 , WO2013/175215 , WO2013/083994 and WO2013/182850 ).
  • any one feature of the compounds of the invention may be combined with any embodiment of another feature of compounds of the invention to create a further embodiment.
  • 'halo' or 'halogen' refers to a fluorine, chlorine, bromine or iodine atom. Particular examples of halo are fluorine and chlorine, especially fluorine.
  • the alkyl group may be straight chain, branched, cyclic, or a combination thereof.
  • Examples of C 1-4 alkyl are methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, cyclopropyl and cyclobutyl.
  • a particular group of exemplary C 1-4 alkyl groups are methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl.
  • An example of C 1-4 alkoxy is methoxy.
  • 'haloC 1-4 alkyl' as used herein, includes straight chain, branched chain or cyclic alkyl groups containing 1 to 4 carbon atoms substituted by one or more halo atoms, for example fluoromethyl, difluoromethyl and trifluoromethyl.
  • a particular group of exemplary haloC 1-4 alkyl include methyl and ethyl groups substituted with one to three halo atoms, in particular one to three fluoro atoms, such as trifluoromethyl or 2,2,2-trifluoroethyl.
  • 'haloC 1-4 alkoxy' as used herein, includes straight chain, branched chain or cyclic alkoxy groups containing 1 to 4 carbon atoms substituted by one or more halo atoms, for example fluoromethoxy, difluoromethoxy and trifluoromethoxy.
  • a particular group of exemplary haloC 1-4 alkyl include methoxy and ethoxy groups substituted with one to three halo atoms, in particular one to three fluoro atoms.
  • the term '5 or 6 membered saturated or unsaturated heterocycle, with at least one O atom' includes for example dihydrofuran, dihydropyran, furan, pyran, oxazole, isoxazole, oxazine, dioxine, morpholine or 1,3-dioxalane.
  • salts of the compounds of formula (I) should be pharmaceutically acceptable. Suitable pharmaceutically acceptable salts will be apparent to those skilled in the art.
  • Pharmaceutically acceptable salts include those described by Berge et al., 1977.
  • Such pharmaceutically acceptable salts include acid addition salts formed with inorganic acids e.g. hydrochloric, hydrobromic, sulphuric, nitric or phosphoric acid and organic acids e.g. succinic, maleic, acetic, fumaric, citric, tartaric, benzoic, p-toluenesulfonic, methanesulfonic or naphthalenesulfonic acid.
  • Other salts e.g. oxalates or formates, may be used, for example in the isolation of compounds of formula (I) and are included within the scope of this invention.
  • Certain of the compounds of formula (I) may form acid addition salts with one or more equivalents of the acid.
  • the present invention includes within its scope all possible stoichiometric and non-stoichiometric forms.
  • the compounds of formula (I) may be prepared in crystalline or non-crystalline form and, if crystalline, may optionally be solvated, e.g. as the hydrate.
  • This invention includes within its scope stoichiometric solvates (e.g. hydrates) as well as compounds containing variable amounts of solvent (e.g. water).
  • isotopes examples include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine, iodine and chlorine such as 2 H (deuterium), 3 H, 11 C, 13 C, 14 C, 18 F, 123 I or 125 I, which may be naturally occurring or non-naturally occurring isotopes. Unless isotopic enrichment is required, suitably the isotope content of the compound of formula (I) is not altered from that commonly found in nature.
  • Isotopically labelled compounds of the present invention for example those into which radioactive isotopes such as 3 H or 14 C have been incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e. 3 H, and carbon-14, i.e. 14 C, isotopes are particularly preferred for their ease of preparation and detectability. 11 C and 18 F isotopes are particularly useful in PET (positron emission tomography).
  • the compounds of formula (I) are intended for use in pharmaceutical compositions it will readily be understood that they are each preferably provided in substantially pure form, for example at least 60% pure, more suitably at least 75% pure and preferably at least 85%, especially at least 98% pure (% are on a weight for weight basis). Impure preparations of the compounds may be used for preparing the more pure forms used in the pharmaceutical compositions.
  • the compounds of formula (I) may be made according to the organic synthesis techniques known to those skilled in this field, as well as by the representative methods set forth below, those in the Examples, and modifications thereof.
  • the present invention provides a modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 channels for use in the prophylaxis or treatment of pain wherein the modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 channels is a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate thereof, as defined above.
  • a 'modulator' as used herein refers to a compound which is capable of producing at least 20% potentiation of whole-cell currents mediated by human Kv3.1 and/or human Kv3.2 and/or human Kv3.3 channels recombinantly expressed in mammalian cells.
  • Compounds of the invention may be tested in an assay such as provided in Example 1 to determine their modulatory properties.
  • the modulator is capable of producing at least 20% potentiation of whole-cell currents mediated by human Kv3.1 channels recombinantly expressed in mammalian cells.
  • the pEC 50 of the modulator is in the range of 4-7 (such as 5-6.5).
  • the modulator is capable of producing at least 20% potentiation of whole-cell currents mediated by human Kv3.2 channels recombinantly expressed in mammalian cells.
  • the pEC 50 of the modulator is in the range of 4-7 (such as 5-6.5).
  • the modulator is capable of producing at least 20% potentiation of whole-cell currents mediated by human Kv3.3 channels recombinantly expressed in mammalian cells.
  • the pEC 50 of the modulator is in the range of 4-7 (such as 5-6.5).
  • the modulator is capable of producing at least 20% potentiation of whole-cell currents mediated by human Kv3.1 and Kv3.2 channels recombinantly expressed in mammalian cells.
  • the modulator is capable of producing at least 20% potentiation of whole-cell currents mediated by human Kv3.1 and Kv3.3 channels recombinantly expressed in mammalian cells.
  • the modulator is capable of producing at least 20% potentiation of whole-cell currents mediated by human Kv3.2 and Kv3.3 channels recombinantly expressed in mammalian cells.
  • the modulator is capable of producing at least 20% potentiation of whole-cell currents mediated by human Kv3.1, Kv3.2 and Kv3.3 channels recombinantly expressed in mammalian cells.
  • the modulator (such as the compound of formula (I) or a pharmaceutically acceptable salt and/or solvate) is selective for modulation of Kv3.1 channels over modulation of Kv3.2 channels.
  • selective it is meant that the modulator demonstrates, for example, at least a 2 fold, 5 fold or 10 fold activity for Kv3.1 channels than for Kv3.2 channels.
  • the activity of a modulator is suitably quantified by its potency as indicated by an EC 50 value.
  • the modulator (such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate) is selective for modulation of Kv3.2 channels over modulation of Kv3.1 channels.
  • the modulator demonstrates, for example at least a 2 fold, 5 fold or 10 fold activity for Kv3.2 channels than for Kv3.1 channels.
  • Compounds of formula (I) or their pharmaceutically acceptable salts and/or solvates wherein W is Wb and R 1 is H may demonstrate greater activity for the Kv3.2 channel over the Kv3.1 channel.
  • Example 15 disclosed in WO2013/175215 is a compound of the invention which demonstrates selectivity for Kv3.2 channels.
  • the modulator (such as the compound of formula (I) or a pharmaceutically acceptable salt and/or solvate) is selective for modulation of Kv3.1 channels over modulation of Kv3.3 channels.
  • selective it is meant that the modulator demonstrates, for example, at least a 2 fold, 5 fold or 10 fold activity for Kv3.1 channels than for Kv3.3 channels.
  • the activity of a modulator is suitably quantified by its potency as indicated by an EC 50 value.
  • the modulator (such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate) is selective for modulation of Kv3.3 channels over modulation of Kv3.1 channels.
  • the modulator demonstrates, for example at least a 2 fold, 5 fold or 10 fold activity for Kv3.3 channels than for Kv3.1 channels.
  • the activity of a modulator is suitably quantified by its potency as indicated by an EC 50 value.
  • the modulator (such as the compound of formula (I) or a pharmaceutically acceptable salt and/or solvate) is selective for modulation of Kv3.2 channels over modulation of Kv3.3 channels.
  • selective it is meant that the modulator demonstrates, for example, at least a 2 fold, 5 fold or 10 fold activity for Kv3.2 channels than for Kv3.3 channels.
  • the activity of a modulator is suitably quantified by its potency as indicated by an EC 50 value.
  • the modulator (such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate) is selective for modulation of Kv3.3 channels over modulation of Kv3.2 channels.
  • the modulator demonstrates, for example at least a 2 fold, 5 fold or 10 fold activity for Kv3.3 channels than for Kv3.2 channels.
  • the activity of a modulator is suitably quantified by its potency as indicated by an EC 50 value.
  • the modulator (such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate) demonstrates comparable activity between modulation of Kv3.1 and Kv3.2 channels, for example the activity for one channel is less than 2 fold that for the other channel, such as less than 1.5 fold or less than 1.2 fold.
  • Compounds of formula (I) or their pharmaceutically acceptable salts and/or solvates wherein W is Wb, R 1 is C 1-4 alkyl, in particular methyl, in the para position may demonstrate comparable activity between modulation of Kv3.1 and Kv3.2 channels.
  • Compound 3 is a compound of the invention which demonstrates a comparable activity between modulation of Kv3.1 and Kv3.2 channels.
  • the activity of a modulator is suitably quantified by its potency as indicated by an EC 50 value.
  • the modulator (such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate) demonstrates comparable activity between modulation of Kv3.1 and Kv3.3 channels, for example the activity for one channel is less than 2 fold that for the other channel, such as less than 1.5 fold or less than 1.2 fold.
  • the activity of a modulator is suitably quantified by its potency as indicated by an EC 50 value.
  • the modulator (such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate) demonstrates comparable activity between modulation of Kv3.2 and Kv3.3 channels, for example the activity for one channel is less than 2 fold that for the other channel, such as less than 1.5 fold or less than 1.2 fold.
  • the activity of a modulator is suitably quantified by its potency as indicated by an EC 50 value.
  • the modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 is selective over other channels which have been associated with pain.
  • the modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 is capable of providing an increase of whole-cell currents of, on average, at least 20% of the increase observed with 50 micromolar N -cyclohexyl- N -[(7,8-dimethyl-2-oxo-1,2-dihydro-3-quinolinyl)methyl]- N '-phenylurea
  • the modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 does not have a notable effect on Kv3.4, Kv7.2/7.3, and/or Nav 1.7 currents.
  • the Examples herein provide suitable methods for the testing of Kv3.1, Kv3.2, Kv3.3, Kv 3.4, Kv7.2/7.3, and Nav 1.7 currents.
  • the modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 is selective over Kv3.4.
  • the modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 is capable of providing an increase of Kv3.1 and/or Kv3.2 and/or Kv3.3 whole-cell currents of, on average, at least 20% of the increase observed with 50 micromolar N -cyclohexyl- N -[(7,8-dimethyl-2-oxo-1,2-dihydro-3-quinolinyl)methyl]- N '-phenylurea but provides an increase of less than 10% (such as less than 5%, especially less than 1% or suitably no increase) in Kv3.4 current at the same concentration (e.g. 10 uM).
  • the modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 is selective over Kv7.2/7.3.
  • the modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 is capable of providing an increase of Kv3.1 and/or Kv3.2 and/or Kv3.3 whole-cell currents of, on average, at least 20% of the increase observed with 50 micromolar N -cyclohexyl- N -[(7,8-dimethyl-2-oxo-1,2-dihydro-3-quinolinyl)methyl]- N '-phenylurea but provides an increase of less than 10% (such as less than 5%, especially less than 1% or suitably no increase) in Kv7.2/7.3 current at the same concentration (e.g. 10 uM).
  • the modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 is selective over Nav1.7.
  • the modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 is capable of providing an increase of Kv3.1 and/or Kv3.2 and/or Kv3.3 whole-cell currents of, on average, at least 20% of the increase observed with 50 micromolar N -cyclohexyl- N -[(7,8-dimethyl-2-oxo-1,2-dihydro-3-quinolinyl)methyl]- N '-phenylurea but provides an increase of less than 10% (such as less than 5%, especially less than 1% or suitably no increase) in Nav1.7 current at the same concentration (e.g. 10 uM).
  • treatment includes the control, mitigation, reduction, or modulation of the disease state or its symptoms.
  • prophylaxis is used herein to mean preventing symptoms of a disease or disorder in a subject or preventing recurrence of symptoms of a disease or disorder in an afflicted subject and is not limited to complete prevention of an affliction.
  • pain that may be mediated by a modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 channels is chronic pain. In another embodiment of the invention, pain is acute pain.
  • the pain indications that may be mediated by a modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 channels is nociceptive, neuropathic, inflammatory or miscellaneous pain.
  • Nociceptive pain represents the normal response to noxious insult or injury of tissues such as skin, muscles, visceral organs, joints, tendons, or bones.
  • Examples of nociceptive pain which form part of the invention include somatic pain: musculoskeletal (joint pain, myofascial pain) or cutaneous, which is often well localized; or visceral pain: hollow organs or smooth muscle.
  • Neuropathic pain is pain initiated or caused by a primary lesion or disease in the somatosensory nervous system. Sensory abnormalities range from deficits perceived as paraesthesia (numbness) to hypersensitivity (hyperalgesia or allodynia), and dysaesthesia (tingling and other sensations). Examples of neuropathic pain which form part of the invention include, but are not limited to, diabetic neuropathy, post-herpetic neuralgia, spinal cord injury pain, phantom limb (post-amputation) pain, and post-stroke central pain. Other causes of neuropathic pain include trauma, chemotherapy and heavy metal exposure.
  • Inflammatory pain occurs as a result of activation and sensitization of the nociceptive pain pathway by a variety of mediators released at a site of tissue inflammation.
  • Mediators that have been implicated as key players in inflammatory pain are pro-inflammatory cytokines such IL-1-alpha, IL-1-beta, IL-6 and TNF-alpha, chemokines, reactive oxygen species, vasoactive amines, lipids, ATP, acid, and other factors released by infiltrating leukocytes, vascular endothelial cells, or tissue resident mast cells.
  • Examples causes of inflammatory pain which form part of the invention include appendicitis, rheumatoid arthritis, inflammatory bowel disease, and herpes zoster.
  • Miscellaneous pain refers to pain conditions or disorders which are not easily classifiable.
  • the current understanding of their underlying mechanisms is still rudimentary though specific therapies for those disorders are well known; they include cancer pain, migraine and other primary headaches and wide-spread pain of the fibromyalgia type.
  • specific pain indications that may be mediated by a modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 channels are neuropathic pain and/or inflammatory pain.
  • the neuropathic pain that may be ameliorated by a modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 channels may be central or peripheral neuropathic pain.
  • Central neuropathic pain is caused by damage to or dysfunction of the central nervous system (CNS), which includes but is not limited to the brain, brainstem, and spinal cord.
  • Peripheral neuropathic pain is caused by damage to or dysfunction of the peripheral nervous system, which includes but is not limited to sensory nerves, motor nerves and autonomic nerves.
  • the neuropathic pain is central neuropathic pain.
  • the neuropathic pain is peripheral neuropathic pain.
  • Pain is a subjective condition and in a clinical setting tends to be measured by a patient's self-assessment. Therefore, it can be difficult to measure and quantify pain threshold.
  • a subjective 11-point rating scale is used where 0 is no pain and 10 is the worst pain imaginable.
  • Subjects generally record their worst pain over a given period, usually a day.
  • a minimum mean baseline score is also recorded and response to the medication is measured relative to the baseline, for example, a reduction of at least 10%, 20%, 30%, 40% or 50% in pain from the baseline score may be observed.
  • a reduction of at least 10%, 20%, 30%, 40% or 50% in pain from the baseline score is observed upon administration of a Kv3.1/Kv3.2/Kv3.3 modulator, such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate thereof to a subject in need thereof.
  • a Kv3.1/Kv3.2/Kv3.3 modulator such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate thereof
  • a Kv3.1/Kv3.2/Kv3.3 modulator can occur before an anticipated onset of pain or after the onset of pain. In cases where it is anticipated that development of a disease or disorder may lead to an increase in pain experienced by the subject, a Kv3.1/Kv3.2/Kv3.3 modulator, such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate thereof can be administered. In cases where a subject is already experiencing pain, a Kv3.1/Kv3.2/Kv3.3 modulator, such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate thereof may be administered to a subject in need thereof.
  • Treatment of the subject in need thereof may continue for as long as treatment is required, for example, 1 day, 1 week, 2 weeks, 3 weeks, 1 month, 6 months, 1 year, more than 1 year more than 2 years, more than 5 years or more than 10 years. Therefore in one embodiment of the invention, a therapeutically effective amount of a Kv3.1/Kv3.2/Kv3.3 modulator, such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate thereof, is administered to a subject in need thereof for 1 day to 1 month, 1 week to 3 months, 1 month to 6 months, 3 months to 1 year or more than 1 year.
  • a Kv3.1/Kv3.2/Kv3.3 modulator such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate thereof
  • Reduction in pain in a subject can be measured by assessing the response to an external stimuli such as mechanical or thermal (e.g. cold) stimuli (such as described in the Experimental section).
  • the reduction can either be considered as a percentage reversal (calculated by measuring the pre- and post-dose thresholds of the affected pain site with a non-affected pain site, such as described in more detail under Data Analysis in the Experimental Section) or by measuring withdrawal thresholds of the affected pain site.
  • the percentage reversal calculation is used.
  • the sensitivity to pain (such as neuropathic pain or inflammatory pain) is reversed by more than 20%, more than 30%, more than 40%, more than 50%, more than 60%, more than 70%, more than 80% or more than 90%, upon administration of a therapeutically effective amount of a Kv3.1/Kv3.2/Kv3.3 modulator, such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate thereof.
  • a Kv3.1/Kv3.2/Kv3.3 modulator such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate thereof.
  • the sensitivity to pain is reversed by more than 80% or more than 90%.
  • Subjects receiving the Kv3.1/Kv3.2/Kv3.3 modulator may experience secondary benefits, such as one or more of improved function, mood, sleep, quality of life, reduced time off work.
  • the prophylaxis or treatment of pain does not include the prophylaxis or treatment of sleep disorder due to neuropathic pain.
  • the prophylaxis or treatment of pain does not include the prophylaxis or treatment of sleep disorder due to pain.
  • the modulators are usually administered as a pharmaceutical composition.
  • the invention also provides a pharmaceutical composition comprising a modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 (such as a compound of formula (I), or a pharmaceutically acceptable salt and/or solvate thereof), and a pharmaceutically acceptable carrier.
  • the modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 may be administered by any convenient method, e.g. by oral, parenteral, buccal, sublingual, nasal, rectal, intrathecal or transdermal administration, and the pharmaceutical compositions adapted accordingly.
  • a modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 which are active when given orally can be formulated as liquids or solids, e.g. as syrups, suspensions, emulsions, tablets, capsules or lozenges.
  • a liquid formulation will generally consist of a suspension or solution of the active ingredient in a suitable liquid carrier(s) e.g. an aqueous solvent such as water, ethanol or glycerine, or a non-aqueous solvent, such as polyethylene glycol or an oil.
  • a suitable liquid carrier(s) e.g. an aqueous solvent such as water, ethanol or glycerine, or a non-aqueous solvent, such as polyethylene glycol or an oil.
  • the formulation may also contain a suspending agent, preservative, flavouring and/or colouring agent.
  • a composition in the form of a tablet can be prepared using any suitable pharmaceutical carrier(s) routinely used for preparing solid formulations, such as magnesium stearate, starch, lactose, sucrose and cellulose.
  • a composition in the form of a capsule can be prepared using routine encapsulation procedures, e.g. pellets containing the active ingredient can be prepared using standard carriers and then filled into a hard gelatin capsule; alternatively a dispersion or suspension can be prepared using any suitable pharmaceutical carrier(s), e.g. aqueous gums, celluloses, silicates or oils and the dispersion or suspension then filled into a soft gelatin capsule.
  • suitable pharmaceutical carrier(s) e.g. aqueous gums, celluloses, silicates or oils
  • Typical parenteral compositions consist of a solution or suspension of the active ingredient in a sterile aqueous carrier or parenterally acceptable oil, e.g. polyethylene glycol, polyvinyl pyrrolidone, lecithin, arachis oil or sesame oil.
  • a sterile aqueous carrier or parenterally acceptable oil e.g. polyethylene glycol, polyvinyl pyrrolidone, lecithin, arachis oil or sesame oil.
  • the solution can be lyophilised and then reconstituted with a suitable solvent just prior to administration.
  • compositions for nasal administration may conveniently be formulated as aerosols, drops, gels and powders.
  • Aerosol formulations typically comprise a solution or fine suspension of the active ingredient in a pharmaceutically acceptable aqueous or non-aqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container which can take the form of a cartridge or refill for use with an atomising device.
  • the sealed container may be a disposable dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve.
  • the dosage form comprises an aerosol dispenser, it will contain a propellant which can be a compressed gas e.g. air, or an organic propellant such as a fluorochlorohydrocarbon or hydrofluorocarbon. Aerosol dosage forms can also take the form of pump-atomisers.
  • compositions suitable for buccal or sublingual administration include tablets, lozenges and pastilles where the active ingredient is formulated with a carrier such as sugar and acacia, tragacanth, or gelatin and glycerin.
  • a carrier such as sugar and acacia, tragacanth, or gelatin and glycerin.
  • compositions for rectal administration are conveniently in the form of suppositories containing a conventional suppository base such as cocoa butter.
  • compositions suitable for transdermal administration include ointments, gels and patches.
  • the composition is in unit dose form such as a tablet, capsule or ampoule.
  • the composition may contain from 0.1% to 100% by weight, for example from 10 to 60% by weight, of the active material, depending on the method of administration.
  • the composition may contain from 0% to 99% by weight, for example 40% to 90% by weight, of the carrier, depending on the method of administration.
  • the composition may contain from 0.05mg to 1000mg, for example from 1.0mg to 500mg, of the active material, depending on the method of administration.
  • the composition may contain from 50 mg to 1000 mg, for example from 100mg to 400mg of the carrier, depending on the method of administration.
  • the dose of the compound used in the treatment of the aforementioned disorders will vary in the usual way with the seriousness of the disorders, the weight of the sufferer, and other similar factors.
  • suitable unit doses may be 0.05 to 1000 mg, more suitably 1.0 to 500mg, and such unit doses may be administered more than once a day, for example two or three a day. Such therapy may extend for a number of weeks or months.
  • the modulator of Kv3.1 and/or Kv3.2 and/or Kv3.3 is used in combination with a further therapeutic agent or agents.
  • the modulators are used in combination with other therapeutic agents, the compounds may be administered either sequentially or simultaneously by any convenient route. Alternatively, the compounds may be administered separately.
  • combinations referred to above may conveniently be presented for use in the form of a pharmaceutical formulation.
  • the individual components of such combinations may be administered either sequentially or simultaneously in separate or combined pharmaceutical formulations.
  • the individual components of combinations may also be administered separately, through the same or different routes.
  • Therapeutic agents which may be used in combination with the present invention include NSAIDS (such as aspirin, naproxen, ibuprofen, parecoxib, diclofenac), paracetamol, pregabalin, gabapentin or opioids (such as fentanyl, sufentanil, oxycodone, morphine, tramadol, codeine).
  • NSAIDS such as aspirin, naproxen, ibuprofen, parecoxib, diclofenac
  • paracetamol pregabalin
  • gabapentin or opioids (such as fentanyl, sufentanil, oxycodone, morphine, tramadol, codeine).
  • Therapeutic agents which may be used in combination for neuropathic pain include pregabalin, duloxetine and capsaicin.
  • Kv3.1/Kv3.2/Kv3.3 modulator such as a compound of formula (I) or a pharmaceutically acceptable salt and/or solvate thereof will suitably be administered at a dosage level which achieves the desired medical outcome without undue adverse effects - namely a safe and effective dose.
  • Example dosages may be in the range of 10mg to 3g per day, such as 200mg to 1.5g per day.
  • a pharmaceutical composition of the invention which may be prepared by admixture, suitably at ambient temperature and atmospheric pressure, is usually adapted for oral, parenteral or rectal administration and, as such, may be in the form of tablets, capsules, oral liquid preparations, powders, granules, lozenges, reconstitutable powders, injectable or infusible solutions or suspensions or suppositories. Orally administrable compositions are generally preferred.
  • the ability of the compounds of the invention to modulate the voltage-gated potassium channel subtypes Kv3.3/Kv3.2/Kv3.1 may be determined using the following assay. Analogous methods may be used to investigate the ability of the compounds of the invention to modulate other channel subtypes.
  • a stable cell line expressing human Kv3.3 channels was created by transfecting Chinese Hamster Ovary (CHO)-K1 cells with a pBacMire_KCNC-3 vector.
  • Cells were cultured in DMEM/F12 (Gibco) supplemented with 10% Foetal Bovine Serum (Gibco), 1X non-essential amino acids (Invitrogen) and geneticin (G418) 400 microg/mL. Cells were grown and maintained at 37°C in a humidified environment containing 5% CO 2 in air.
  • hKv3.2 a stable cell line expressing human Kv3.2 channels (hKv3.2) was created by transfecting CHO-K1 cells with a pCIH5-hKv3.2 vector.
  • Cells were cultured in DMEM/F12 medium supplemented by 10% Foetal Bovine Serum, 1X non-essential amino acids (Invitrogen) and 100ug/ml of Hygromycin-B (Invitrogen). Cells were grown and maintained at 37°C in a humidified environment containing 5% CO 2 in air.
  • CHO/Gam/E1A-clone22 alias CGE22 cells were transduced using a hKv3.1 BacMam reagent.
  • This cell line was designed to be an improved CHO-K1-based host for enhanced recombinant protein expression as compared to wild type CHO-K1.
  • the cell line was generated following the transduction of CHO-K1 cells with a BacMam virus expressing the Adenovirus-Gam1 protein and selection with Geneticin-G418, to generate a stable cell line, CHO/Gam-A3.
  • CHO/Gam-A3 cells were transfected with pCDNA3-E1A-Hygro, followed by hygromycin-B selection and FACS sorting to obtain single-cell clones.
  • BacMam-Luciferase and BacMam-GFP viruses were then used in transient transduction studies to select the clone based on highest BacMam transduction and recombinant protein expression.
  • CGE22 cells were cultured in the same medium used for the hKv3.2 CHO-K1 stable cell line with the addition of 300ug/ml hygromycin-B and 300ug/ml G418. All other conditions were identical to those for hKv3.2 CHO-K1 cells.
  • Leak subtraction was conducted in all experiments by applying 50 ms hyperpolarizing (10 mV) prepulses to evoke leak currents followed by a 20 ms period at the holding potential before test pulses.
  • 50 ms hyperpolarizing (10 mV) prepulses to evoke leak currents followed by a 20 ms period at the holding potential before test pulses.
  • a first test pulse to -15 mV was applied for 100 ms and following a further 100 ms at -70 mV, a second pulse to 40 mV was applied for 50 ms.
  • Cells were then maintained for a further 100 ms at -100 mV and then a voltage ramp from -100 mV to 40 mV was applied over 200 ms.
  • a first test pulse to 0 mV was applied for 500 ms and following a further 100 ms at -70 mV, a second pulse to 40 mV was applied for 200 ms. These longer test pulses were used to study inactivation of hKv3.3 channels.
  • Test pulses protocol may be performed in the absence (pre-read) and presence (post-read) of the test compound. Pre- and post-reads may be separated by the compound addition followed by a 3-minute incubation.
  • the intracellular solution contained the following (in mM): K-gluconate 100, KCI 54, MgCl 2 3.2, HEPES 5, adjusted to pH 7.3 with KOH.
  • Amphotericin-B solution was prepared as 50mg/ml stock solution in DMSO and diluted to a final working concentration of 0.1 mg/ml in intracellular solution.
  • the external solution was Dulbecco's Phosphate Buffered Saline (DPBS) and contained the following (in mM): CaCl 2 0.90, KCI 2.67, KH 2 PO 4 1.47, MgCl.6H 2 O 0.493, NaCl 136.9, Na 3 PO 4 8.06, with a pH of 7.4.
  • the recordings were analysed and filtered using both seal resistance (>20 M ⁇ ) and peak current amplitude (>500pA at the voltage step of 40 mV) in the absence of compound to eliminate unsuitable cells from further analysis.
  • seal resistance >20 M ⁇
  • peak current amplitude >500pA at the voltage step of 40 mV
  • paired comparisons of evoked currents between pre- and post-drug additions measured for the -15 mV voltage step were used to determine the positive modulation effect of each compound.
  • Kv3 channel-mediated outward currents were measured determined from the mean amplitude of the current over the final 10ms of the -15mV voltage pulse minus the mean baseline current at -70mV over a 10ms period just prior to the -15mV step.
  • Kv3 channel currents following addition of the test compound were then compared with the currents recorded prior to compound addition.
  • Data were normalised to the maximum effect of the reference compound (50microM of N -cyclohexyl- N -[(7,8-dimethyl-2-oxo-1,2-dihydro-3-quinolinyl)methyl]- N '-phenylurea) and to the effect of a vehicle control (0.5% DMSO).
  • the normalised data were analysed using ActivityBase or Excel software.
  • the concentration of compound required to increase currents by 50% of the maximum increase produced by the reference compound (EC 50 ) was determined by fitting of the concentration-response data using a four parameter logistic function in ActivityBase.
  • paired comparisons of evoked currents between pre- and post-drug additions were measured for the 0mV step, considering the peak current and the decay (inactivation) of the current over the duration of the 0mv test pulse (500ms).
  • N -cyclohexyl- N -[(7,8-dimethyl-2-oxo-1,2-dihydro-3-quinolinyl)methyl]- N '-phenylurea was obtained from ASINEX (Registry Number: 552311-06-5).
  • Kv3.1 and/or Kv3.2 positive modulators produce in the above assay an increase of whole-cell currents of, on average, at least 20% of the increase observed with 50 micromolar N -cyclohexyl- N -[(7,8-dimethyl-2-oxo-1,2-dihydro-3-quinolinyl)methyl]- N '-phenylurea.
  • Compound 1 was found to have a pEC50 for Kv3.3 of 5.17.
  • Compound 2 was found to have a pEC50 for Kv3.3 of 4.93.
  • Compound 3 was found to have a pEC50 for Kv3.3 of 4.76.
  • a secondary analysis of the data from the hKv3.1, hKv3.2 and hKv3.3 assays described in Example 1 may be used to investigate the effect of the compounds on rate of rise of the current from the start of the depolarising voltage pulses.
  • the magnitude of the effect of a compound can be determined from the time constant (Tau act ) obtained from a non-linear fit, using the equation given below, of the rise in Kv3.1, Kv3.2 and Kv3.3 currents following the start of the -15mV depolarising voltage pulse.
  • Tau act time constant
  • K is the rate constant
  • Tau act is the activation time constant, which is the reciprocal of K.
  • the effect of the compounds on the time taken for Kv3.1, Kv3.2 or Kv3.3 currents to decay on closing of the channels at the end of the -15mV depolarising voltage pulses can also be investigated.
  • the magnitude of the effect of a compound on channel closing can be determined from the time constant (Tau deact ) of a non-linear fit of the decay of the current ("tail current") immediately following the end of the depolarising voltage pulse.
  • Kv3.1, Kv3.2 and Kv3.3 channels must activate and deactivate very rapidly in order to allow neurons to fire actions potentials at high frequency (Rudy et al., 2001). Slowing of activation is likely to delay the onset of action potential repolarisation; slowing of deactivation could lead to hyperpolarising currents that reduce the excitability of the neuron and delay the time before the neuron can fire a further action potential. Together these two slowing effects on channel activation and deactivation are likely to lead to a reduction rather than a facilitation of the neurons ability to fire at high frequencies.
  • Subjects comprised male, Wistar Hanover rats, 6 animals per group (225 ⁇ 2g for Compound 1 studies and Compound 2 studies; 214 ⁇ 1g for Compound 3 studies; 236 ⁇ 1g for Compound 4 studies).
  • Vehicle 12% Captisol ® ; 0.5% w/v HPMC and 0.1% w/v Tween-80; 5 ml/kg via the intraperitoneal route
  • Vehicle 12% Captisol ® ; 0.5% w/v HPMC and 0.1% w/v Tween-80; 5 ml/kg via the intraperitoneal route
  • Table 1 Dosing regimen for neuropathic and inflammatory pain models.
  • Compound Neuropathic Pain Inflammatory Pain Study 1 Study 2 Compound 1 30 mg/kg (i.p.) 1 10 mg/kg (i.p.) 10 mg/kg (i.p.) 60 mg/kg (i.p.) 30 mg/kg (i.p.) 30 mg/kg (i.p.) 60 mg/kg (i.p.) 60 mg/kg (i.p.) Compound 2 30 mg/kg (i.p.) 10 mg/kg (i.p.) 10 mg/kg (i.p.) 60 mg/kg (i.p.) 30 mg/kg (i.p.) 30 mg/kg (i.p.) 60 mg/kg (i.p.) 60 mg/kg (i.p.) Compound 3 30 mg/kg (i.p.) N/A 10 mg/kg (i.p.) 60 mg/kg (i.p.) 30 mg/kg (i.p.) 30 mg/kg (i.p
  • the control for the neuropathic pain model was lamotrigine, administered at 30 mg/kg via oral delivery.
  • the control for the inflammatory pain model was diclofenac, administered at 30 mg/kg via oral delivery.
  • Statistical analysis was performed using one-way ANOVA, and comparisons were performed with time-matched vehicle group using Tukey's HSD test wherein * p ⁇ 0.05, ** p ⁇ 0.01, *** p ⁇ 0.001.
  • Treatment groups were randomised and blinded. Groups of 6 rats were used.
  • Neuropathic pain was induced by partial ligation of the sciatic nerve. Briefly, the rats were anaesthetised (isoflurane/O 2 inhalation), the left sciatic nerve exposed at mid-thigh level through a small incision and 1/3 to 1/2 of the nerve thickness tightly ligated within a 7.0 silk suture. The wound was closed with surgical glue. Animals were allowed to recover and tested 12-15 days following surgery.
  • Withdrawal thresholds or latencies were measured on both the ipsilateral (ligated) and contralateral (non-ligated) paws, prior to (predose) and then up to 24 h following drug or vehicle administration.
  • Pre-dose behavioural measurements were obtained by measuring paw withdrawals 14 days following nerve ligation; before the initiation of drug treatment. Following treatment, further readings were taken at 1, 3, 6 and 24 hour after administration.
  • the hyperalgesia was induced by an intraplantar injection (25 ⁇ l) of Freund's Complete Adjuvant (FCA) into the left hind paw.
  • FCA Freund's Complete Adjuvant
  • paw withdrawal thresholds or latencies were measured on both the ipsilateral (FCA-injected) and contralateral (non-injected) paws, prior to (na ⁇ ve) and 24 hours following FCA injection (predose), and then at 1, 3, 6 and 24 hours after drug or vehicle administration.
  • Cold sensitivity was assessed using a commercially available cold-plate (Ugo Basile, Milan). The cold plate was allowed to stabilize for 5 minutes at the set temperature prior to testing. Paw withdrawal latencies (PWL) were determined with the cold-plate set at 10°C. The animals were lightly restrained and each hind paw in turn placed onto the surface of the cold-plate. The end point was taken as the withdrawal of the paw and recorded as the withdrawal latency for the ipsilateral and the contralateral paw. A maximum cut-off of 30 seconds was used for each paw.
  • PWL Paw withdrawal latencies
  • % reversal ipsilaterd threshold postdose ⁇ ipsilaterd threshold predose contralateral threshold predose ⁇ ipsilaterd threshold predose ⁇ 100
  • % reversal left postdose PWT / L ⁇ left predose PWT / L left na ⁇ ve PWT / L ⁇ left predose PWT / L ⁇ 100
  • Partial ligation of the sciatic nerve resulted in a marked decrease in withdrawal threshold to a mechanical stimulus and in withdrawal latency to a cold stimulus of the affected paw.
  • predose threshold readings of 66 ⁇ 1g were measured in the ipsilateral paws compared to 106 ⁇ 1g in the contralateral paws ( Fig.1a, Fig 1b ).
  • Cold latencies of 7.0 ⁇ 0.2s were measured in the ipsilateral paws compared to 10.9 ⁇ 0.2s in the contralateral paws ( Fig.2a , Fig 2b ).
  • Compound 1 produced a reversal of both mechanical ( Fig.1a , Fig.1c ) and cold sensitivity ( Fig.2a , Fig.2c ) with rapid onset of action and good dose separation.
  • Peak reversal of mechanical sensitivity was seen at 3 hours post-dose (51% at 30mg/kg and 68% by 60mg/kg). Cold sensitivity was reversed at 3 hours post-dose by 52% and 125% by 30 mg/kg and 60mg/kg respectively.
  • the lower dose of the compound was only statistically active against mechanical hyperalgesia. At 60mg/kg the compound was still efficacious at 6 hours post-dose.
  • the positive control, lamotrigine gave peak reversals of 65% (at 3 hours post-dose) and 58% (at 1 hour post-dose) in mechanical and cold respectively.
  • Partial ligation of the sciatic nerve resulted in a marked decrease in withdrawal threshold to a mechanical stimulus and in withdrawal latency to a cold stimulus of the affected paw.
  • predose threshold readings of 66 ⁇ 1g were measured in the ipsilateral paws compared to 104 ⁇ 1g in the contralateral paws ( Fig.3a, Fig.3b ).
  • Cold latencies of 6.8 ⁇ 0.1s were measured in the ipsilateral paws compared to 10.8 ⁇ 0.2s in the contralateral paws ( Fig.4a , Fig.4b ).
  • Compound 1 produced a dose-related reversal of mechanical ( Fig.3a , Fig.3c ) and cold sensitivity ( Fig.4a , Fig.4c ) with rapid onset and long duration of action.
  • Peak reversal of mechanical sensitivity was seen at 3 hours post-dose (31% at 10mg/kg, 73% at 30mg/kg and 81% by 60mg/kg). Cold sensitivity was reversed at 3 hours post-dose by 39%, 68% and 76% by 10, 30 and 60mg/kg respectively.
  • FCA intraplantar injection of FCA resulted in a marked decrease in withdrawal threshold to a mechanical stimulus and in withdrawal latency to a cold stimulus of the affected paw.
  • the mean na ⁇ ve threshold readings were 105 ⁇ 1g.
  • predose threshold readings of 65 ⁇ 1.0g were measured in the ipsilateral paws compared to 104 ⁇ 1.0g in the contralateral paws ( Fig.5a, Fig.5b ).
  • the mean naive cold latency readings were 11.8 ⁇ 0.2s.
  • Compound 1 produced a dose-related reversal of both mechanical ( Fig.5a , Fig.5c ) and cold sensitivity ( Fig.6a , Fig.6b ) with rapid onset of action and peak reversal at 1-3 h post-dose.
  • Peak reversal of mechanical sensitivity was seen at 1 hour post-dose for 30mg/kg and 60mg/kg (74% and 92% respectively) and at 3 hours post-dose for 10mg/kg (64% reversal).
  • Peak reversal of cold sensitivity was seen at 3 hours post-dose for 10mg/kg and 30mg/kg (45% and 65% respectively) and at 1 hour post-dose for 60mg/kg (92% reversal).
  • Partial ligation of the sciatic nerve resulted in a marked decrease in withdrawal threshold to a mechanical stimulus and in withdrawal latency to a cold stimulus of the affected paw.
  • predose threshold readings of 67 ⁇ 1g were measured in the ipsilateral paws compared to 106 ⁇ 1g in the contralateral paws ( Fig.7a, Fig.7b ).
  • Cold latencies of 7.0 ⁇ 0.2s were measured in the ipsilateral paws compared to 11.3 ⁇ 0.3s in the contralateral paws ( Fig.8a , Fig.8b ).
  • Compound 2 produced a reversal of both mechanical ( Fig.7a , Fig.7c ) and cold sensitivity ( Fig.8a , Fig.8c ) with rapid onset of action and efficacy similar to lamotrigine.
  • Partial ligation of the sciatic nerve resulted in a marked decrease in withdrawal threshold to a mechanical stimulus and in withdrawal latency to a cold stimulus of the affected paw.
  • predose threshold readings of 66 ⁇ 1g were measured in the ipsilateral paws compared to 104 ⁇ 1g in the contralateral paws ( Fig.9a, Fig.9b ).
  • Cold latencies of 6.8 ⁇ 0.1s were measured in the ipsilateral paws compared to 10.8 ⁇ 0.2s in the contralateral paws ( Fig.10a , Fig.10b ).
  • Compound 2 produced a dose-related reversal of mechanical ( Fig.9a , Fig.9c ) and cold sensitivity ( Fig.10a , Fig.10c ) with rapid onset of action.
  • Peak reversal of mechanical sensitivity was seen at 3 hours post-dose (31% at 10mg/kg, 72% at 30mg/kg and 81% by 60mg/kg). Cold sensitivity was reversed at 3 hours post-dose by 35%, 70% and 95% by 10, 30 and 60mg/kg respectively.
  • FCA intraplantar injection of FCA resulted in a marked decrease in withdrawal threshold to a mechanical stimulus and in withdrawal latency to a cold stimulus of the affected paw.
  • the mean na ⁇ ve threshold readings were 106 ⁇ 1g. Twenty-four hours after FCA injection, predose threshold readings of 67 ⁇ 1.0g were measured in the ipsilateral paws compared to 106 ⁇ 1.0g in the contralateral paws ( Fig.11a, Fig.11b ). The mean naive cold latency readings were 11.1 ⁇ 0.2s.
  • Compound 2 produced a dose-related reversal of both mechanical ( Fig.11a , Fig.11c ) and cold sensitivity ( Fig.12a , Fig.12c ) with rapid onset of action and peak reversal at 1-3 h post-dose.
  • Peak reversal of mechanical sensitivity was seen at 3 hours post-dose (46% at 10 mg/kg and 67% at 60mg/kg). Peak reversal of cold sensitivity was seen at 1 hour post-dose (96% with 60mg/kg), and seen at 3 hours post-dose for 30 mg/kg (58% reversal). At 3 hours post-dose, the compound was still efficacious at 60 mg/kg (81% reversal).
  • Partial ligation of the sciatic nerve resulted in a marked decrease in withdrawal threshold to a mechanical stimulus and in withdrawal latency to a cold stimulus of the affected paw.
  • predose threshold readings 64 ⁇ 1g were measured in the ipsilateral paws compared to 104 ⁇ 1g in the contralateral paws ( Fig.13a, Fig.13b ).
  • Cold latencies of 7.0 ⁇ 0.2s were measured in the ipsilateral paws compared to 11.2 ⁇ 0.2s in the contralateral paws ( Fig.14a , Fig.14b ).
  • Compound 3 produced a dose-related reversal of both mechanical ( Fig.13a , Fig.13c ) and cold sensitivity ( Fig.14a , Fig.14c ) with slow onset of action.
  • FCA The intraplantar injection of FCA resulted in a marked decrease in withdrawal threshold to a mechanical stimulus and in withdrawal latency to a cold stimulus of the affected paw.
  • example Compound 3 produced a reversal of either mechanical ( Fig.15a , Fig.15c ) or cold sensitivity ( Fig.16a , Fig.16c ).
  • the compound had a slow onset of action, with little reversal evident at 1 hour post-dose. Peak reversal was seen at 3 h post-dose for both mechanical and cold sensitivity after which it declined but activity still evident at 6 hours post-dose (significant on mechanical thresholds).
  • FCA The intraplantar injection of FCA resulted in a marked decrease in withdrawal threshold to a mechanical stimulus and in withdrawal latency to a cold stimulus of the affected paw.
  • the mean naive threshold readings were 104 ⁇ 1g.
  • predose threshold readings of 64 ⁇ 1.0g were measured in the ipsilateral paws compared to 104 ⁇ 1.0g in the contralateral paws ( Fig.17a, Fig 17b ).
  • the mean naive cold latency readings were 11.1 ⁇ 0.1s.
  • Compound 4 produced a reversal of the mechanical ( Fig.17a , Fig.17c ) and cold hyperalgesia ( Fig.18a , Fig.18c ) that was largely dose-related.
  • the compound had peak reversal at 1-3 h post-dose after which it declined, but significant activity still evident at 6h post-dose.
  • Example 3 Specificity of compounds as potentiators of Kv3.1 and/or 3.2 and/or 3.3
  • intracellular solution was loaded into the intracellular compartments of the QPlate and cell suspension was pipetted into the extracellular compartments.
  • membrane currents were recorded using up to 48 parallel patch clamp amplifiers in the QPatch HT ® system. The current records were sampled at 2000 Hz and low-pass Bessel filtered at 400 Hz.
  • Human Kv3.4 channels were stably expressed in a HEK293 cell line.
  • Cells were used within 1-4 hours of cell preparation and internal and external physiological solutions were freshly prepared prior to the assay. Electrophysiological recordings were made using an automated patch clamp platform (QPatch, Sophion Biosciences).
  • the extracellular solution contained 145 mM NaCl, 4 mM KCI, 2mM CaCl 2 , 1 mM MgCl 2 , 10 mM HEPES and 10 mM glucose; the pH was adjusted to 7.4 with NaOH, and the osmolarity measured as 313 mOsm/L.
  • a low potassium intracellular solution which contained 135.6 mM CsCI, 5.37 mM CaCl 2 , 1.75 mM MgCl 2 , 10 mM EGTA, 15.6 mM KOH, 4 mM Na 2 ATP and 10 mM HEPES; the pH was adjusted to 7.2 with CsOH, and the osmolarity measured as 303 mOsm/L.
  • the voltage dependence of inactivation of the homomeric hKv3.4 channels was tested using a protocol that consisted of a series of two square voltage pulses.
  • Pulse one varied between the holding potential of -60mV to +40mV and pulse two stepped from a holding potential of -60 to +40mV before returning to the holding potential of -60mV.
  • the time constants of the inactivation phase of the hKv3.4 currents were derived from a mono ( ⁇ ) or a double ( ⁇ 1 and ⁇ 2 ) standard exponential fit to peak trace pulse from -80mV to +60mV.
  • the time-course of effect of the compounds against the hKv3.4 potassium channel was measured with repetitive voltage pulses from a holding potential of -60mV, with pulses of 500ms duration every 5s to +40mV.
  • test compounds concentration (10 uM) of the test compounds was assessed during the activation, inactivation and peak pulse protocols. The protocol was applied with addition of physiological solution (control period), followed by 10 uM of the testing compound and a 10 mM of the standard blocker TEA.
  • Stock solutions (10mM) of the standard blockers TEA was prepared in fresh physiological solution prior to testing on the e-phys automated platform.
  • the test compounds were prepared in 10mM DMSO stock and then diluted 1 in 1000 in physiological solution.
  • hKv3.4 channels were heterologously expressed in Xenopus ooctyes following microinjection of in vitro transcribed mRNA (mMessage mMachine kit, Ambion, Austin, TX).
  • Whole-oocyte currents were recorded 1-4 day post-microinjection at room temperature (21-23°C) under two-electrode voltage-clamp conditions (OC-725C, Warner, Hamden, CT).
  • ND-96 and ND-96 plus the test compound were delivered using a gravity-driven perfusion system.
  • manual patch clamp was carried out using an Axon 200B amplifier (Axon Instruments).
  • the software program pCIamp (version 10) from Axon Instruments was used to stimulate and record electrical activity.
  • GraphPad Prism (version 5) software was used to analyse the data.
  • steady-state voltage pulses (1 s) were applied every 10 s from a holding potential of -80 mV to +60 mV, in 10 mV steps. Following a control period of at least 3 min, compounds were perfused for at least 3 min. The compounds were dissolved in 100% DMSO (at 10 mM) and subsequently diluted in a physiological solution without exceeding 0.1% DMSO in a final concentration of 10 ⁇ M. Concentration of 0.1% DMSO did not lead to significant effects on the amplitude of the fully activated peak of the hKv7.2/7.3 channel, which generally remain stable for the duration of a typical whole-cell recording.
  • Perfusion of the test compounds was started if cells had less than 10% run-down within a two full I-V control protocols. Cells with higher run-down were excluded from further analysis. Following perfusion of test compounds, agonist retigabine (from Alomone Labs, Israel) and blocker TEA (from Sigma) were tested in the same cells. Retigabine and TEA were prepared as 10 mM and 100 mM stock solution in DMSO and water, respectively. Stock solutions were subsequently diluted in physiological solutions at the final concentrations of 10 ⁇ M and 10 mM, respectively.
  • Human Nav1.7 channels were stably expressed in a HEK293 cell line. Electrophysiological recordings were made at room temperature using an automated patch clamp platform (QPatch, Sophion Biosciences).
  • the QPatch assay was carried out at room temperature using the QPatch platform (Sophion). On the day of the experiment cells were cultured according to standard cell preparation for QPatch. Internal and external physiological solutions were freshly prepared prior to the assay.
  • the standard extracellular solution contained 145mM NaCl, 4mM KCI, 2mM CaCl 2 , 2mM MgCl 2 , 10mM HEPES and 10mM glucose; the pH was adjusted to 7.4 with NaOH, and the osmolarity measured as 314mOsm/L.
  • the standard intracellular solution contained 140mM CsF, 10mM NaCl, 5mM CsOH, 1mM EGTA, 10mM HEPES; the pH was adjusted to 7.25 with CsOH, and the osmolarity measured as 293mOsm/L.
  • a series of 40 voltage pulses (from -120mV to 0mV, 2.4 ms long at 117Hz) were applied every 60s in control (twice), compound (five times), standard blocker (twice) and washout (twice).
  • the % inhibition values were calculated by normalising the data relative to the 1st pulse (P1) and by calculating the ratio between the 40th pulse versus the 1st pulse (P40/1). Series resistance and quality of seals were monitored during the experiments. Sophion QPatch Assay Software 5.0 was used to analyse and plot all the graphs. 10 ⁇ M of the test compound was applied followed by 1 ⁇ M TTX (standard blocker), followed by saline (washout).
  • Seizure Beget Seizure The Quest for GABA as a Key Player. Crit. Rev. Neurobiol. 2006;18(1-2):135-144 .
  • Fisahn A Kainate receptors and rhythmic activity in neuronal networks: hippocampal gamma oscillations as a tool. J. Physiol. 2005 Oct;561(1):65-72 .

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Pain & Pain Management (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Rheumatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Furan Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
EP16813020.1A 2015-12-10 2016-12-09 Modulators of kv3 channels to treat pain Active EP3386506B1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1521751.6A GB201521751D0 (en) 2015-12-10 2015-12-10 Novel uses
PCT/GB2016/053879 WO2017098254A1 (en) 2015-12-10 2016-12-09 Modulators of kv3 channels to treat pain

Publications (2)

Publication Number Publication Date
EP3386506A1 EP3386506A1 (en) 2018-10-17
EP3386506B1 true EP3386506B1 (en) 2024-01-24

Family

ID=55274489

Family Applications (1)

Application Number Title Priority Date Filing Date
EP16813020.1A Active EP3386506B1 (en) 2015-12-10 2016-12-09 Modulators of kv3 channels to treat pain

Country Status (17)

Country Link
US (2) US11147813B2 (zh)
EP (1) EP3386506B1 (zh)
JP (2) JP7149847B2 (zh)
KR (1) KR20180097549A (zh)
CN (1) CN108472288B (zh)
AU (1) AU2016367239B2 (zh)
BR (1) BR112018011700B1 (zh)
CA (1) CA3005302A1 (zh)
DK (1) DK3386506T3 (zh)
EA (1) EA038059B1 (zh)
FI (1) FI3386506T3 (zh)
GB (1) GB201521751D0 (zh)
IL (1) IL259717B (zh)
MX (1) MX2018007074A (zh)
PT (1) PT3386506T (zh)
SG (1) SG11201804123VA (zh)
WO (1) WO2017098254A1 (zh)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201521751D0 (en) 2015-12-10 2016-01-27 Autifony Therapeutics Ltd Novel uses
GB201613163D0 (en) 2016-07-29 2016-09-14 Autifony Therapeutics Ltd Novel compounds
WO2018109484A1 (en) 2016-12-16 2018-06-21 Autifony Therapeutics Limited Hydantoin modulators of kv3 channels
WO2018220762A1 (ja) * 2017-05-31 2018-12-06 大塚製薬株式会社 ピリミジン化合物
BR112021006940A2 (pt) * 2018-10-16 2021-07-13 Autifony Therapeutics Limited compostos
AR116898A1 (es) * 2018-10-30 2021-06-23 H Lundbeck As DERIVADOS DE ARILSULFONILPIRROLCARBOXAMIDA COMO ACTIVADORES DE CANALES DE POTASIO Kv3
KR20220139923A (ko) 2020-02-06 2022-10-17 오티포니 세라피틱스 리미티드 Kv3 조절제
CN117751119A (zh) 2021-08-10 2024-03-22 奥蒂福尼疗法有限公司 钾通道调节剂
CN113788741B (zh) * 2021-09-28 2023-12-29 大连九信精细化工有限公司 一种制备2-环丙基苯酚衍生物的方法

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110098300A1 (en) 2008-06-06 2011-04-28 Ucb Pharma, S.A. Compounds Comprising A Cyclobutoxy Group
CA2781685C (en) * 2009-12-11 2018-09-04 Autifony Therapeutics Limited Imidazolidinedione derivatives
PL2649066T3 (pl) * 2010-12-06 2017-04-28 Autifony Therapeutics Limited Pochodne hydantoiny przydatne jako inhibitory KV3
CN103596943B (zh) 2011-06-07 2016-10-12 奥蒂福尼疗法有限公司 用作kv3抑制剂的乙内酰脲衍生物
GB201209986D0 (en) 2012-06-06 2012-07-18 Autifony Therapeutics Ltd Novel compounds
DK2788339T3 (en) * 2011-12-06 2016-05-23 Autifony Therapeutics Ltd HYDANTOIN DERIVATIVES, USEFUL AS QU3 INHIBITORS
BR112014028718A2 (pt) 2012-05-22 2017-06-27 Autifony Therapeutics Ltd derivados de hidantoína como inibidores de kv3
WO2013175215A1 (en) * 2012-05-22 2013-11-28 Autifony Therapeutics Limited Triazoles as kv3 inhibitors
WO2013182851A1 (en) 2012-06-06 2013-12-12 Autifony Therapeutics Limited Prophylaxis or treatment of diseases where a modulator of kv3.3 channels is required
GB201521751D0 (en) 2015-12-10 2016-01-27 Autifony Therapeutics Ltd Novel uses
GB201522179D0 (en) 2015-12-16 2016-01-27 Autifony Therapeutics Ltd Novel compounds

Also Published As

Publication number Publication date
DK3386506T3 (da) 2024-05-06
KR20180097549A (ko) 2018-08-31
EA201891377A1 (ru) 2019-01-31
IL259717A (en) 2018-07-31
SG11201804123VA (en) 2018-06-28
FI3386506T3 (fi) 2024-04-23
WO2017098254A1 (en) 2017-06-15
US11944623B2 (en) 2024-04-02
JP2022141699A (ja) 2022-09-29
CA3005302A1 (en) 2017-06-15
JP2019506367A (ja) 2019-03-07
US11147813B2 (en) 2021-10-19
CN108472288B (zh) 2024-05-28
AU2016367239B2 (en) 2020-07-09
EP3386506A1 (en) 2018-10-17
IL259717B (en) 2021-05-31
BR112018011700A2 (pt) 2018-11-27
BR112018011700B1 (pt) 2023-12-26
US20190000848A1 (en) 2019-01-03
CN108472288A (zh) 2018-08-31
EA038059B1 (ru) 2021-06-30
GB201521751D0 (en) 2016-01-27
US20220071998A1 (en) 2022-03-10
PT3386506T (pt) 2024-03-28
MX2018007074A (es) 2018-12-12
AU2016367239A1 (en) 2018-06-07
JP7149847B2 (ja) 2022-10-07

Similar Documents

Publication Publication Date Title
EP3386506B1 (en) Modulators of kv3 channels to treat pain
EP3390394B1 (en) Hydantoin modulators of kv3 channels
US8637510B2 (en) Morpholinothiazoles as alpha 7 positive allosteric modulators
Iftinca Neuronal T–type calcium channels: What's new? Iftinca: T–type channel regulation
WO2013182851A1 (en) Prophylaxis or treatment of diseases where a modulator of kv3.3 channels is required
EP2852588B1 (en) Hydantoin derivatives as kv3 inhibitors
CN104080782B (zh) 用作激酶活性调节剂的咪唑胺
HRP20050601A2 (en) Chk-, pdk-, and akt-inhibitory pyrimidines, their production and use as pharmarmaceutical agents
EP3490977B1 (en) Cyclobutane derivatives as modulators of voltage-gated potassium channels
CN101631546A (zh) 螺-吲哚酮化合物作为治疗剂的用途
JP2008542219A (ja) 疼痛処置法
Suter et al. Rufinamide attenuates mechanical allodynia in a model of neuropathic pain in the mouse and stabilizes voltage-gated sodium channel inactivated state
EA034244B1 (ru) 1-гетероарил-индолин-4-карбоксамиды в качестве модуляторов gpr52, применяемые для лечения или предотвращения расстройств, связанных с gpr52
JP2018529679A (ja) ピラゾリル置換ヘテロアリール及び医薬としてのその使用
WO2018109484A1 (en) Hydantoin modulators of kv3 channels
Cascio et al. In vitro and in vivo pharmacological characterization of two novel selective cannabinoid CB2 receptor inverse agonists
Murineddu et al. Different classes of CB2 ligands potentially useful in the treatment of pain
Spasov et al. Novel Approaches to the Development of Antimigraine Drugs: A Focus on 5-HT 2A Receptor Antagonists
TW202031645A (zh) 作為Kv3鉀通道激活劑之芳基磺醯基吡咯甲醯胺衍生物
WO2019075394A1 (en) COMPOUNDS FOR THE TREATMENT OF EMOTIONAL / PSYCHOLOGICAL SYMPTOMS IN FRAGILE X SYNDROME

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20180614

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1262362

Country of ref document: HK

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20200318

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

RAP3 Party data changed (applicant data changed or rights of an application transferred)

Owner name: AUTIFONY THERAPEUTICS LIMITED

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

RIC1 Information provided on ipc code assigned before grant

Ipc: C07D 307/77 20060101ALI20230703BHEP

Ipc: A61P 29/00 20060101ALI20230703BHEP

Ipc: A61K 31/506 20060101ALI20230703BHEP

Ipc: A61K 31/4439 20060101ALI20230703BHEP

Ipc: A61K 31/4196 20060101AFI20230703BHEP

INTG Intention to grant announced

Effective date: 20230801

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602016085495

Country of ref document: DE

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20240206

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Ref document number: 3386506

Country of ref document: PT

Date of ref document: 20240328

Kind code of ref document: T

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20240325

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20240503

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG9D